EP3762031A1 - Anticorps anti-claudine 18.2 et leurs utilisations - Google Patents

Anticorps anti-claudine 18.2 et leurs utilisations

Info

Publication number
EP3762031A1
EP3762031A1 EP19764603.7A EP19764603A EP3762031A1 EP 3762031 A1 EP3762031 A1 EP 3762031A1 EP 19764603 A EP19764603 A EP 19764603A EP 3762031 A1 EP3762031 A1 EP 3762031A1
Authority
EP
European Patent Office
Prior art keywords
seq
chain variable
variable region
polypeptide sequence
light chain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP19764603.7A
Other languages
German (de)
English (en)
Other versions
EP3762031A4 (fr
Inventor
Minghan Wang
Hui Zou
Haiqun JIA
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Phanes Therapeutics Inc
Original Assignee
Phanes Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Phanes Therapeutics Inc filed Critical Phanes Therapeutics Inc
Publication of EP3762031A1 publication Critical patent/EP3762031A1/fr
Publication of EP3762031A4 publication Critical patent/EP3762031A4/fr
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • G01N33/6857Antibody fragments
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • This invention relates to monoclonal anti-claudin 18.2 (CLDN18.2) antibodies, nucleic acids and expression vectors encoding the antibodies, recombinant cells containing the vectors, and compositions comprising the antibodies. Methods of making the antibodies, and methods of using the antibodies to treat diseases including cancer and inflammatory diseases and/or associated complications are also provided.
  • CLDN18.2 monoclonal anti-claudin 18.2
  • This application contains a sequence listing, which is submitted electronically via EFS-Web as an ASCII formatted sequence listing with a file name“689204.9WO
  • Claudin 18.2 (CLDN18.2), also known as claudin-l8a2.
  • CLDN belongs to the claudin (CLDN) family transmembrane proteins of at least 27 isoforms in humans.
  • Claudins are the major structural components of tight junction between epithelial cells and function as ion pores to regulate the paracellular permeability of cations and anions (Sahin et ah, Physiol Rev. 2013; 93 :525-569).
  • the expression of CLDN18 is normally limited to lung and stomach tissues.
  • CLDN18 has two splicing variants.
  • CLDN18.1 is the lung-specific variant whereas CLDN18.2 is the stomach-specific variant.
  • the splicing variants differ at their N-terminal 69 amino acid residues due to alternative splicing of the first exon (Niimi et ah, Mol Cell Biol. 2001; 21 :7380-7390).
  • CLDN18.2 knockout mice suggest that CLDN18.2 plays a critical role in preventing gastric acid leakage into the stomach lumen (Hayash et al., Gastroenterology 2012; 142:292-304).
  • CLDN18.2 is an ideal target for precision-guided, anti-cancer biologies to
  • CLDN18.2-positive tumors as CLDN18.2 is not expressed in any normal tissue other than the differentiated gastric epithelial cells, which are unreachable by large molecule compounds administrated intravenously.
  • the invention relates to isolated monoclonal antibodies or antigen-binding fragments thereof that specifically bind claudin 18.2 (CLDN18.2).
  • HCDR1 heavy chain complementarity determining region 1
  • HCDR2 heavy chain complementarity determining region 1
  • LCDR3 light chain complementarity determining region 1
  • HCDR1 heavy chain complementarity determining region 1
  • HCDR2 heavy chain complementarity determining region 1
  • LCDR3 light chain complementarity determining region 1
  • the antibody or antigen-binding fragment thereof specifically binds CLDN18.2, preferably human CLDN18.2.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 9, 1, 3, 5, 7, 11, 13, 15, 17 or 19, or a light chain variable region having a polypeptide sequence at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 10, 2, 4, 6, 8, 12, 14, 16, 18 or 20.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises:
  • the isolated monoclonal antibody or antigen-binding fragment thereof binds to CLDN18.2 and is capable of inducing effector-mediated tumor cell lysis through antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent phagocytosis (ADPC), and/or complement-dependent cytotoxicity (CDC), and/or mediating the recruitment of conjugated drugs, and/or forming a bispecific antibody with another monoclonal antibody or antigen-binding fragment thereof with cancer-killing effect.
  • ADCC antibody-dependent cellular cytotoxicity
  • ADPC antibody-dependent phagocytosis
  • CDC complement-dependent cytotoxicity
  • the isolated monoclonal antibody or antigen-binding fragment thereof is chimeric.
  • the isolated monoclonal antibody or antigen-binding fragment thereof is human or humanized.
  • the humanized monoclonal antibody or antigen-binding fragment thereof comprises:
  • isolated nucleic acids encoding the monoclonal antibodies or antigen-binding fragments thereof of the invention.
  • vectors comprising the isolated nucleic acids encoding the monoclonal antibodies or antigen-binding fragments thereof of the invention.
  • host cells comprising the vectors comprising the isolated nucleic acids encoding the monoclonal antibodies or antigen-binding fragments thereof of the invention.
  • composition comprising an isolated monoclonal antibody or antigen-binding fragment thereof of the invention and a pharmaceutically acceptable carrier.
  • CLDN18.1 on a cancer cell surface in a subject in need thereof, comprising
  • the cancer can be any liquid or solid cancer, for example, it can be selected from, but not limited to, a lung cancer, a gastric cancer, an esophageal cancer, a bile duct cancer, a cholangiocarcinoma, a colon cancer, a hepatocellular carcinoma, a renal cell carcinoma, a bladder urothelial carcinoma, a metastatic melanoma, a breast cancer, an ovarian cancer, a cervical cancer, a head and neck cancer, a pancreatic cancer, a glioma, a glioblastoma, and other solid tumors, and a non-Hodgkin’s lymphoma (NHL), an acute lymphocytic leukemia (ALL), a chronic lymphocytic leukemia (CLL), a chronic myelogenous leukemia (CML),
  • NHL non-Hodgkin’s lymphoma
  • ALL acute lymphocytic leukemia
  • CLL chronic lympho
  • Also provided are methods of producing a pharmaceutical composition comprising a monoclonal antibody or antigen-binding fragment thereof of the invention, comprising combining the monoclonal antibody or antigen-binding fragment thereof with a pharmaceutically acceptable carrier to obtain the pharmaceutical composition.
  • kits for determining the level of CLDN18.2 in a subject comprise (a) obtaining a sample from the subject; (b) contacting the sample with an antibody or antigen-binding fragment thereof of the invention; and (c) determining the level of CLDN18.2 in the subject.
  • the sample is a tissue sample.
  • the tissue sample can, for example, be a cancer tissue sample.
  • the sample is a blood sample.
  • FIGs. 1 A-1D show the dose-dependent binding of purified chimeric anti- CLDN18.2 mAbs (VH and VL regions of mouse mAbs fused to the constant regions of human IgGl heavy chain and kappa light chain, respectively) to a HEK293 cell pool stably transfected with full-length human CLDN18.2 by FACS analysis.
  • FIGs. 2A-2E show the selective binding of chimeric anti-CLDNl8.2 mAbs to CLDN18.2 compared with CLDN18.1.
  • the shaded gray peak is representative of the treatment group incubated with only the secondary antibody and the open black peak is representative of the treatment group incubated with both primary chimeric mAh and the secondary antibody.
  • FIGs. 3 A-3C show the results of dose-dependent binding of the chimeric anti- CLDN18.2 mAbs to a HEK293-CLDN18.2 stable cell line by FACS analysis.
  • FIGs. 4A-4J show the histograms for the binding of the chimeric anti- CLDN18.2 mAbs to a HEK293-CLDN18.2 stable cell line.
  • the shaded gray peak is representative of the treatment group incubated with only the secondary antibody and the open black peak is representative of the treatment group incubated with both primary chimeric mAh and the secondary antibody.
  • FIGs. 5A-5G show the results of dose-dependent binding of the humanized anti- CLDN18.2 mAbs to HEK293-CLDN18.2 stable pools by FACS analysis.
  • FIGs. 6A-6E show the results of dose-dependent binding of the humanized anti- CLDN18.2 mAbs to a HEK293-CLDN18.2 stable cell line by FACS analysis.
  • FIG. 7 shows the binding of the humanized anti-CLDNl8.2 mAbs to the cancer cell line NUGC-4 at mAh concentration of 685 nM by FACS analysis.
  • FIGs. 8A-8B show the results for the antibody-dependent cellular cytotoxicity (ADCC) activity of five humanized anti-CLDNl8.2 mAbs and one chimeric anti- CLDN18.2 mAh.
  • ADCC antibody-dependent cellular cytotoxicity
  • any numerical values such as a concentration or a concentration range described herein, are to be understood as being modified in all instances by the term“about.”
  • a numerical value typically includes ⁇ 10% of the recited value.
  • a concentration of 1 mg/mL includes 0.9 mg/mL to 1.1 mg/mL.
  • a concentration range of 1% to 10% (w/v) includes 0.9% (w/v) to 11% (w/v).
  • the use of a numerical range expressly includes all possible subranges, all individual numerical values within that range, including integers within such ranges and fractions of the values unless the context clearly indicates otherwise.
  • the terms“comprises,”“comprising,”“includes,”“including,” “has,”“having,”“contains” or“containing,” or any other variation thereof will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers and are intended to be non-exclusive or open-ended.
  • a composition, a mixture, a process, a method, an article, or an apparatus that comprises a list of elements is not necessarily limited to only those elements but can include other elements not expressly listed or inherent to such composition, mixture, process, method, article, or apparatus.
  • “or” refers to an inclusive or and not to an exclusive or. For example, a condition A or B is satisfied by any one of the following: A is true (or present) and B is false (or not present), A is false (or not present) and B is true (or present), and both A and B are true (or present).
  • the conjunctive term“and/or” between multiple recited elements is understood as encompassing both individual and combined options. For instance, where two elements are conjoined by“and/or,” a first option refers to the applicability of the first element without the second. A second option refers to the applicability of the second element without the first. A third option refers to the applicability of the first and second elements together. Any one of these options is understood to fall within the meaning, and therefore satisfy the requirement of the term“and/or” as used herein.
  • “subject” means any animal, preferably a mammal, most preferably a human.
  • the term“mammal” as used herein, encompasses any mammal. Examples of mammals include, but are not limited to, cows, horses, sheep, pigs, cats, dogs, mice, rats, rabbits, guinea pigs, monkeys, humans, etc., more preferably a human.
  • nucleic acids or polypeptide sequences e.g., anti-CLDNl8.2 antibodies and amino acids
  • polynucleotides that encode them refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same, when compared and aligned for maximum correspondence, as measured using one of the following sequence comparison algorithms or by visual inspection.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are input into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated.
  • sequence comparison algorithm then calculates the percent sequence identity for the test sequence(s) relative to the reference sequence, based on the designated program parameters.
  • Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol. 48:443 (1970), by the search for similarity method of Pearson & Lipman, Proc. NatT. Acad. Sci. USA 85:2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by visual inspection (see generally, Current Protocols in Molecular Biology, F.M. Ausubel et ak, eds., Current Protocols, a joint venture between Greene Publishing Associates, Inc. and John Wiley & Sons, Inc., (1995 Supplement) (Ausubel)).
  • BLAST and BLAST 2.0 algorithms are described in Altschul et al. (1990) J. Mol. Biol. 215: 403-410 and Altschul et al. (1997) Nucleic Acids Res. 25: 3389-3402, respectively.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information.
  • This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al, supra).
  • HSPs high scoring sequence pairs
  • T is referred to as the neighborhood word score threshold (Altschul et al, supra).
  • These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them.
  • the word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased.
  • the BLASTP program uses as defaults a wordlength (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff & Henikoff, Proc. Natl. Acad. Sci. USA 89: 10915 (1989)).
  • the BLAST algorithm In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin & Altschul, Proc. NatT. Acad. Sci. USA 90:5873-5787 (1993)).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
  • a further indication that two nucleic acid sequences or polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the polypeptide encoded by the second nucleic acid, as described below.
  • a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative
  • nucleic acid sequences are substantially identical is that the two molecules hybridize to each other under stringent conditions.
  • the invention generally relates to isolated anti-CLDNl8.2 antibodies, nucleic acids and expression vectors encoding the antibodies, recombinant cells containing the vectors, and compositions comprising the antibodies. Methods of making the antibodies, and methods of using the antibodies to treat diseases including cancer and inflammatory diseases.
  • the antibodies of the invention possess one or more desirable functional properties, including but not limited to high-affinity binding to CLDN18.2, high specificity to CLDN18.2, the ability to stimulate complement-dependent cytotoxicity (CDC), antibody-dependent phagocytosis (ADPC), and/or antibody-dependent cellular- mediated cytotoxicity (ADCC) against cells expressing CLDN18.2, and the ability to inhibit tumor growth in subjects and animal models when administered alone or in combination with other anti -cancer therapies.
  • CDC complement-dependent cytotoxicity
  • ADPC antibody-dependent phagocytosis
  • ADCC antibody-dependent cellular- mediated cytotoxicity
  • the term“antibody” is used in a broad sense and includes immunoglobulin or antibody molecules including human, humanized, composite and chimeric antibodies and antibody fragments that are monoclonal or polyclonal. In general, antibodies are proteins or peptide chains that exhibit binding specificity to a specific antigen. Antibody structures are well known. Immunoglobulins can be assigned to five major classes (i.e., IgA, IgD, IgE, IgG and IgM), depending on the heavy chain constant domain amino acid sequence. IgA and IgG are further sub-classified as the isotypes IgAl, IgA2, IgGl, IgG2, IgG3 and IgG4. Accordingly, the antibodies of the invention can be of any of the five major classes or corresponding sub-classes.
  • the antibodies of the invention are IgGl, IgG2, IgG3 or IgG4.
  • Antibody light chains of vertebrate species can be assigned to one of two clearly distinct types, namely kappa and lambda, based on the amino acid sequences of their constant domains.
  • the antibodies of the invention can contain a kappa or lambda light chain constant domain.
  • the antibodies of the invention include heavy and/or light chain constant regions from rat or human antibodies.
  • antibodies contain an antigen-binding region that is made up of a light chain variable region and a heavy chain variable region, each of which contains three domains (i.e., complementarity determining regions 1-3; CDR1, CDR2, and CDR3).
  • the light chain variable region domains are alternatively referred to as LCDR1, LCDR2, and LCDR3, and the heavy chain variable region domains are alternatively referred to as HCDR1, HCDR2, and HCDR3.
  • an“isolated antibody” refers to an antibody which is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds to CLDN18.2 is substantially free of antibodies that do not bind to CLDN18.2). In addition, an isolated antibody is substantially free of other cellular material and/or chemicals.
  • the term“monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts.
  • the monoclonal antibodies of the invention can be made by the hybridoma method, phage display technology, single lymphocyte gene cloning technology, or by recombinant DNA methods.
  • the monoclonal antibodies can be produced by a hybridoma which includes a B cell obtained from a transgenic nonhuman animal, such as a transgenic mouse or rat, having a genome comprising a human heavy chain transgene and a light chain transgene.
  • the term“antigen-binding fragment” refers to an antibody fragment such as, for example, a diabody, a Fab, a Fab', a F(ab')2, an Fv fragment, a disulfide stabilized Fv fragment (dsFv), a (dsFv) 2 , a bispecific dsFv (dsFv-dsFv 1 ), a disulfide stabilized diabody (ds diabody), a single-chain antibody molecule (scFv), a single domain antibody (sdab) an scFv dimer (bivalent diabody), a multispecific antibody formed from a portion of an antibody comprising one or more CDRs, a camelized single domain antibody, a nanobody, a domain antibody, a bivalent domain antibody, or any other antibody fragment that binds to an antigen but does not comprise a complete antibody structure.
  • an antibody fragment such as, for example, a diabody, a Fab
  • an antigen-binding fragment is capable of binding to the same antigen to which the parent antibody or a parent antibody fragment binds.
  • the antigen-binding fragment comprises a light chain variable region, a light chain constant region, and an Fd segment of the heavy chain.
  • the antigen-binding fragment comprises Fab and F(ab’).
  • the term“single-chain antibody” refers to a conventional single chain antibody in the field, which comprises a heavy chain variable region and a light chain variable region connected by a short peptide of about 15 to about 20 amino acids.
  • the term“single domain antibody” refers to a conventional single domain antibody in the field, which comprises a heavy chain variable region and a heavy chain constant region or which comprises only a heavy chain variable region.
  • human antibody refers to an antibody produced by a human or an antibody having an amino acid sequence corresponding to an antibody produced by a human made using any technique known in the art. This definition of a human antibody includes intact or full-length antibodies, fragments thereof, and/or antibodies comprising at least one human heavy and/or light chain polypeptide.
  • humanized antibody refers to a non-human antibody that is modified to increase the sequence homology to that of a human antibody, such that the antigen-binding properties of the antibody are retained, but its antigenicity in the human body is reduced.
  • chimeric antibody refers to an antibody wherein the amino acid sequence of the immunoglobulin molecule is derived from two or more species.
  • the variable region of both the light and heavy chains often corresponds to the variable region of an antibody derived from one species of mammal (e.g., mouse, rat, rabbit, etc.) having the desired specificity, affinity, and capability, while the constant regions correspond to the sequences of an antibody derived from another species of mammal (e.g., human) to avoid eliciting an immune response in that species.
  • the term“multispecific antibody” refers to an antibody that comprises a plurality of immunoglobulin variable domain sequences, wherein a first immunoglobulin variable domain sequence of the plurality has binding specificity for a first epitope and a second immunoglobulin variable domain sequence of the plurality has binding specificity for a second epitope.
  • the first and second epitopes are on the same antigen, e.g., the same protein (or subunit of a multimeric protein).
  • the first and second epitopes overlap or substantially overlap.
  • the first and second epitopes do not overlap or do not substantially overlap.
  • the first and second epitopes are on different antigens, e.g. , different proteins (or different subunits of a multimeric protein).
  • a first immunoglobulin variable domain sequence of the plurality has binding specificity for a first epitope
  • a second immunoglobulin variable domain sequence of the plurality has binding specificity for a second epitope.
  • multispecific antibody comprises a third, fourth, or fifth immunoglobulin variable domain.
  • a multispecific antibody is a bispecific antibody molecule, a trispecific antibody molecule, or a tetraspecific antibody molecule.
  • the term“bispecifc antibody” refers to a multispecific antibody that binds no more than two epitopes or two antigens.
  • a bispecific antibody is characterized by a first immunoglobulin variable domain sequence which has binding specificity for a first epitope and a second immunoglobulin variable domain sequence that has binding specificity for a second epitope.
  • the first and second epitopes are on the same antigen, e.g. , the same protein (or subunit of a multimeric protein).
  • the first and second epitopes overlap or substantially overlap.
  • the first and second epitopes are on different antigens, e.g.
  • a bispecific antibody comprises a heavy chain variable domain sequence and a light chain variable domain sequence which have binding specificity for a first epitope and a heavy chain variable domain sequence and a light chain variable domain sequence which have binding specificity for a second epitope.
  • a bispecific antibody comprises a half antibody, or fragment thereof, having binding specificity for a first epitope and a half antibody, or fragment thereof, having binding specificity for a second epitope.
  • a bispecific antibody comprises a scFv, or fragment thereof, having binding specificity for a first epitope, and a scFv, or fragment thereof, having binding specificity for a second epitope.
  • the first epitope is located on CLDN18.2 and the second epitope is located on PD-l, PD-L1, TIM-3, LAG-3, CTLA-4, EGFR, HER-2, CD 19, CD20, CD33, CD3, CD73, CD47, TPM, apelin, DLL3, folate receptor alpha, and/or other tumor associated immune suppressors or surface antigens.
  • CLDN18.2 refers to claudin 18 variant 2, claudin- 18.2 or claudin- l8a2. l, which belongs to the claudin family of transmembrane proteins. CLDN18.2 is specifically expressed on the surface of epithelial cells in stomach (Niimi et al., Mol Cell Biol. 2001; 21 :7380-7390) and becomes one of the major structural components of the tight junction between the epithelial cells (Sahin et al., Physiol Rev. 2013; 93 :525-569).
  • the term“human CLDN18.2” refers to a CLDN18.2 originated from a human. An exemplary amino acid sequence of a human CLDN18.2 is represented in GenBank Accession No. AAL15637.1 (SEQ ID NO: 141).
  • an antibody that“specifically binds to CLDN18.2” refers to an antibody that binds to a CLDN18.2, preferably a human CLDN18.2, with a KD of 1 c 10 -7 M or less, preferably 1 c 10 -8 M or less, more preferably 5x 10 -9 M or less, 1 c 10 -9 M or less, 5 c 10 -10 M or less, or 1 c 10 -10 M or less.
  • KD refers to the dissociation constant, which is obtained from the ratio of Kd to Ka (i.e., Kd/Ka) and is expressed as a molar concentration (M).
  • KD values for antibodies can be determined using methods in the art in view of the present disclosure.
  • the KD of an antibody can be determined by using surface plasmon resonance, such as by using a biosensor system, e.g., a Biacore® system, or by using bio-layer interferometry technology, such as an Octet RED96 system.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof comprising a heavy chain complementarity determining region 1 (HCDR1), a HCDR2, a HCDR3, a light chain complementarity determining region 1 (LCDR1), a LCDR2, and a LCDR3, having the polypeptide sequences of:
  • the antibody or antigen-binding fragment thereof specifically binds CLDN18.2, preferably human CLDN18.2.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof comprising a heavy chain complementarity determining region 1 (HCDR1), a HCDR2, a HCDR3, a light chain complementarity determining region 1 (LCDR1), a LCDR2, and a LCDR3, having the polypeptide sequences of:
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof comprising a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to one of SEQ ID NOs: 9, 1, 3, 5, 7, 11, 13, 15, 17 or 19, or a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to one of SEQ ID NOs: 10, 2, 4, 6, 8, 12, 14, 16, 18 or 20.
  • the isolated monoclonal antibody or antigen-binding fragment thereof of the invention comprises a heavy chain variable region having the polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 9, 1, 3, 5, 7, 11, 13, 15, 17 or 19, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 10, 2, 4, 6, 8, 12, 14, 16, 18 or 20, respectively.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof of the invention, comprising:
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 21, 22, 23, 51, 52 and 53, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: l, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:2.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 1; and a light chain variable region having the polypeptide sequence of SEQ ID NO:2.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 24, 25, 26, 54, 55 and 56, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:3, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:4.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO:3; and a light chain variable region having the polypeptide sequence of SEQ ID NO:4.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 27, 28, 29, 57, 58 and 59, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:5, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:6.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO:5; and a light chain variable region having the polypeptide sequence of SEQ ID NO:6.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 30, 31, 32, 60, 61 and 62, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 7, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 8.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO:7; and a light chain variable region having the polypeptide sequence of SEQ ID NO: 8.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 33, 34, 35, 63, 64 and 65, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 9, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 10.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO:9; and a light chain variable region having the polypeptide sequence of SEQ ID NO: 10.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 36, 37, 38, 66, 67 and 68, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 11, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 12.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 11; and a light chain variable region having the polypeptide sequence of SEQ ID NO: 12.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 39, 40, 41, 69, 70 and 71, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 13, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 14.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 13; and a light chain variable region having the polypeptide sequence of SEQ ID NO: 14.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 42, 43, 44, 72, 73 and 74, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 15, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 16.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 15; and a light chain variable region having the polypeptide sequence of SEQ ID NO: 16.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 45, 46, 47, 75, 76 and 77, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 17, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 18.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 17; and a light chain variable region having the polypeptide sequence of SEQ ID NO: 18.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 48, 49, 50, 78, 79 and 80, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 19, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:20.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 19; and a light chain variable region having the polypeptide sequence of SEQ ID NO:20.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 81, 82, 83, 111,
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: l, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:2.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 1; and a light chain variable region having the polypeptide sequence of SEQ ID NO:2.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 84, 85, 86, 114, 115 and 116, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:3, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:4.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO:3; and a light chain variable region having the polypeptide sequence of SEQ ID NO:4.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 87, 88, 89, 117, 118 and 119, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:5, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:6.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO:5; and a light chain variable region having the polypeptide sequence of SEQ ID NO:6.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 90, 91, 92, 120, 121 and 122, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 7, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 8.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO:7; and a light chain variable region having the polypeptide sequence of SEQ ID NO: 8.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 93, 94, 95, 123, 124 and 125, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 9, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 10.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO:9; and a light chain variable region having the polypeptide sequence of SEQ ID NO: 10.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 96, 97, 98, 126, 127 and 128, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 11, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 12.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 11; and a light chain variable region having the polypeptide sequence of SEQ ID NO: 12.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 99, 100, 101, 129, 130 and 131, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 13, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 14.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 13; and a light chain variable region having the polypeptide sequence of SEQ ID NO: 14.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 102, 103, 104, 132, 133 and 134, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 15, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 16.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 15; and a light chain variable region having the polypeptide sequence of SEQ ID NO: l6.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 105, 106, 107, 135, 136 and 137, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 17, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 18.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 17; and a light chain variable region having the polypeptide sequence of SEQ ID NO: l8.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof, comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, having the polypeptide sequences of SEQ ID NOs: 108, 109, 110, 138, 139 and 140, respectively.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO: 19, and a light chain variable region having a polypeptide sequence at least 85%, preferably 90%, more preferably 95% or more, such as 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:20.
  • the isolated monoclonal antibody or antigen-binding fragment thereof comprises a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 19; and a light chain variable region having the polypeptide sequence of SEQ ID NO:20.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof of the invention, wherein the antibody or antigen-binding fragment thereof is chimeric.
  • the invention relates to an isolated monoclonal antibody or antigen-binding fragment thereof of the invention, wherein the antibody or antigen-binding fragment thereof is human or humanized.
  • the invention relates to an isolated humanized monoclonal antibody or antigen-binding fragment thereof, wherein the isolated humanized antibody or antigen-binding fragment thereof comprises:
  • n a heavy chain variable region having the polypeptide sequence of SEQ ID NO: 155, and a light chain variable region having the polypeptide sequence of SEQ ID NO: 157;
  • the invention in another general aspect, relates to an isolated nucleic acid encoding a monoclonal antibody or antigen-binding fragment thereof of the invention. It will be appreciated by those skilled in the art that the coding sequence of a protein can be changed (e.g., replaced, deleted, inserted, etc.) without changing the amino acid sequence of the protein. Accordingly, it will be understood by those skilled in the art that nucleic acid sequences encoding monoclonal antibodies or antigen-binding fragments thereof of the invention can be altered without changing the amino acid sequences of the proteins. [0097] In another general aspect, the invention relates to a vector comprising an isolated nucleic acid encoding a monoclonal antibody or antigen-binding fragment thereof of the invention.
  • the vector is a recombinant expression vector such as a plasmid.
  • the vector can include any element to establish a conventional function of an expression vector, for example, a promoter, ribosome binding element, terminator, enhancer, selection marker, and origin of replication.
  • the promoter can be a constitutive, inducible, or repressible promoter.
  • a number of expression vectors capable of delivering nucleic acids to a cell are known in the art and can be used herein for production of an antibody or antigen -binding fragment thereof in the cell. Conventional cloning techniques or artificial gene synthesis can be used to generate a recombinant expression vector according to embodiments of the invention.
  • the invention in another general aspect, relates to a host cell comprising an isolated nucleic acid encoding a monoclonal antibody or antigen-binding fragment thereof of the invention.
  • a host cell comprising an isolated nucleic acid encoding a monoclonal antibody or antigen-binding fragment thereof of the invention.
  • Any host cell known to those skilled in the art in view of the present disclosure can be used for recombinant expression of antibodies or antigen binding fragments thereof of the invention.
  • the host cells are E. coli TG1 or BL21 cells (for expression of, e.g., an scFv or Fab antibody), CHO-DG44 or CHO-K1 cells or HEK293 cells (for expression of, e.g., a full-length IgG antibody).
  • the recombinant expression vector is transformed into host cells by conventional methods such as chemical transfection, heat shock, or electroporation, where it is stably integrated into the host cell genome such that the recombinant nucleic acid is effectively expressed.
  • the invention in another general aspect, relates to a method of producing a monoclonal antibody or antigen-binding fragment thereof of the invention, comprising culturing a cell comprising a nucleic acid encoding the monoclonal antibody or antigen binding fragment thereof under conditions to produce a monoclonal antibody or antigen binding fragment thereof of the invention, and recovering the antibody or antigen-binding fragment thereof from the cell or cell culture (e.g., from the supernatant).
  • Expressed antibodies or antigen-binding fragments thereof can be harvested from the cells and purified according to conventional techniques known in the art and as described herein.
  • the invention in another general aspect, relates to a pharmaceutical composition, comprising an isolated monoclonal antibody or antigen-binding fragment thereof of the invention and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprising an isolated monoclonal antibody or antigen-binding fragment thereof of the invention and a pharmaceutically acceptable carrier.
  • “pharmaceutical composition” as used herein means a product comprising an antibody of the invention together with a pharmaceutically acceptable carrier. Antibodies of the invention and compositions comprising them are also useful in the manufacture of a medicament for therapeutic applications mentioned herein.
  • the term“carrier” refers to any excipient, diluent, filler, salt, buffer, stabilizer, solubilizer, oil, lipid, lipid containing vesicle, microsphere, liposomal encapsulation, or other material well known in the art for use in pharmaceutical formulations. It will be understood that the characteristics of the carrier, excipient or diluent will depend on the route of administration for a particular application.
  • the term“pharmaceutically acceptable carrier” refers to a non-toxic material that does not interfere with the effectiveness of a composition according to the invention or the biological activity of a composition according to the invention. According to particular embodiments, in view of the present disclosure, any pharmaceutically acceptable carrier suitable for use in an antibody pharmaceutical composition can be used in the invention.
  • compositions of the invention are known in the art, e.g., Remington: The Science and Practice of Pharmacy (e.g. 2lst edition (2005), and any later editions).
  • additional ingredients include: buffers, diluents, solvents, tonicity regulating agents, preservatives, stabilizers, and chelating agents.
  • One or more pharmaceutically acceptable carrier can be used in formulating the pharmaceutical compositions of the invention.
  • the pharmaceutical composition is a liquid formulation.
  • a preferred example of a liquid formulation is an aqueous formulation, i.e., a formulation comprising water.
  • the liquid formulation can comprise a solution, a suspension, an emulsion, a microemulsion, a gel, and the like.
  • An aqueous formulation typically comprises at least 50% w/w water, or at least 60%, 70%, 75%,
  • the pharmaceutical composition can be formulated as an injectable which can be injected, for example, via an injection device (e.g., a syringe or an infusion pump).
  • the injection can be delivered subcutaneously, intramuscularly, intraperitoneally, intravitreally, or intravenously, for example.
  • the pharmaceutical composition is a solid formulation, e.g., a freeze-dried or spray-dried composition, which can be used as is, or whereto the physician or the patient adds solvents, and/or diluents prior to use.
  • Solid dosage forms can include tablets, such as compressed tablets, and/or coated tablets, and capsules (e.g., hard or soft gelatin capsules).
  • the pharmaceutical composition can also be in the form of sachets, dragees, powders, granules, lozenges, or powders for reconstitution, for example.
  • the dosage forms may be immediate release, in which case they can comprise a water-soluble or dispersible carrier, or they can be delayed release, sustained release, or modified release, in which case they can comprise water-insoluble polymers that regulate the rate of dissolution of the dosage form in the gastrointestinal tract or under the skin.
  • the pharmaceutical composition can be delivered intranasally, intrabuccally, or sublingually.
  • the pH in an aqueous formulation can be between pH 3 and pH 10.
  • the pH of the formulation is from about 7.0 to about 9.5. In another embodiment of the invention, the pH of the formulation is from about 3.0 to about 7.0.
  • the pharmaceutical composition comprises a buffer.
  • buffers include: arginine, aspartic acid, bicine, citrate, disodium hydrogen phosphate, fumaric acid, glycine, glycylglycine, histidine, lysine, maleic acid, malic acid, sodium acetate, sodium carbonate, sodium dihydrogen phosphate, sodium phosphate, succinate, tartaric acid, tricine, and
  • the buffer can be present individually or in the aggregate, in a concentration from about 0.01 mg/ml to about 50 mg/ml, for example from about 0.1 mg/ml to about 20 mg/ml.
  • Pharmaceutical compositions comprising each one of these specific buffers constitute alternative embodiments of the invention.
  • the pharmaceutical composition comprises a preservative.
  • preservatives include: benzethonium chloride, benzoic acid, benzyl alcohol, bronopol, butyl 4-hydroxybenzoate,
  • the preservative can be present individually or in the aggregate, in a concentration from about 0.01 mg/ml to about 50 mg/ml, for example from about 0.1 mg/ml to about 20 mg/ml. Pharmaceutical compositions comprising each one of these specific preservatives constitute alternative embodiments of the invention.
  • the pharmaceutical composition comprises an isotonic agent.
  • an isotonic agent such as sodium chloride
  • an amino acid such as glycine, histidine, arginine, lysine, isoleucine, aspartic acid, tryptophan, and threonine
  • an alditol such as glycerol, 1,2- propanediol propyleneglycol), 1,3 -propanediol, and l,3-butanediol
  • polyethyleneglycol e.g. PEG400
  • Another example of an isotonic agent includes a sugar.
  • Non-limiting examples of sugars may be mono-, di-, or polysaccharides, or water- soluble glucans, including for example fructose, glucose, mannose, sorbose, xylose, maltose, lactose, sucrose, trehalose, dextran, pullulan, dextrin, cyclodextrin, alpha and beta-HPCD, soluble starch, hydroxyethyl starch, and sodium carboxymethylcellulose.
  • Another example of an isotonic agent is a sugar alcohol, wherein the term“sugar alcohol” is defined as a C(4-8) hydrocarbon having at least one -OH group.
  • Non-limiting examples of sugar alcohols include mannitol, sorbitol, inositol, galactitol, dulcitol, xylitol, and arabitol.
  • the isotonic agent can be present individually or in the aggregate, in a concentration from about 0.01 mg/ml to about 50 mg/ml, for example from about 0.1 mg/ml to about 20 mg/ml.
  • Pharmaceutical compositions comprising each one of these specific isotonic agents constitute alternative embodiments of the invention.
  • the pharmaceutical composition comprises a chelating agent.
  • chelating agents include citric acid, aspartic acid, salts of ethylenediaminetetraacetic acid (EDTA), and mixtures thereof.
  • the chelating agent can be present individually or in the aggregate, in a concentration from about 0.01 mg/ml to about 50 mg/ml, for example from about 0.1 mg/ml to about 20 mg/ml.
  • Pharmaceutical compositions comprising each one of these specific chelating agents constitute alternative embodiments of the invention.
  • the pharmaceutical composition comprises a stabilizer.
  • stabilizers include one or more aggregation inhibitors, one or more oxidation inhibitors, one or more surfactants, and/or one or more protease inhibitors.
  • the pharmaceutical composition comprises a stabilizer, wherein said stabilizer is carboxy-/hydroxycellulose and derivates thereof (such as HPC, HPC-SL, HPC-L and HPMC), cyclodextrins, 2-methylthioethanol, polyethylene glycol (such as PEG 3350), polyvinyl alcohol (PVA), polyvinyl
  • the stabilizer can be present individually or in the aggregate, in a concentration from about 0.01 mg/ml to about 50 mg/ml, for example from about 0.1 mg/ml to about 20 mg/ml.
  • Pharmaceutical compositions comprising each one of these specific stabilizers constitute alternative embodiments of the invention.
  • the pharmaceutical composition comprises one or more surfactants, preferably a surfactant, at least one surfactant, or two different surfactants.
  • surfactant refers to any molecules or ions that are comprised of a water-soluble (hydrophilic) part, and a fat-soluble (lipophilic) part.
  • the surfactant can, for example, be selected from the group consisting of anionic surfactants, cationic surfactants, nonionic surfactants, and/or zwitterionic surfactants.
  • the surfactant can be present individually or in the aggregate, in a concentration from about 0.1 mg/ml to about 20 mg/ml. Pharmaceutical compositions comprising each one of these specific surfactants constitute alternative embodiments of the invention.
  • the pharmaceutical composition comprises one or more protease inhibitors, such as, e.g., EDTA, and/or benzamidine hydrochloric acid (HC1).
  • the protease inhibitor can be present individually or in the aggregate, in a concentration from about 0.1 mg/ml to about 20 mg/ml.
  • Pharmaceutical compositions comprising each one of these specific protease inhibitors constitute alternative embodiments of the invention.
  • the invention in another general aspect, relates to a method of producing a pharmaceutical composition comprising a monoclonal antibody or antigen-binding fragment thereof of the invention, comprising combining a monoclonal antibody or antigen-binding fragment thereof with a pharmaceutically acceptable carrier to obtain the pharmaceutical composition.
  • the invention in another general aspect, relates to a method of targeting CLDN18.2 on a cancer cell surface in a subject to achieve cell killing, the method comprising administering to the subject an isolated monoclonal antibody or antigen binding fragment thereof that specifically binds CLDN18.2 or a pharmaceutical composition comprising the isolated monoclonal antibody or antigen binding fragment thereof of the invention.
  • Binding of the CLDN18.2 monoclonal antibody or antigen binding fragment to CLDN18.2 can mediate complement-dependent cytotoxicity (CDC), antibody-dependent phagocytosis (ADPC), and/or antibody-dependent cellular cytotoxicity (ADCC) or other effects that result in the death of the targeted cancer cell.
  • the monoclonal antibody or antigen binding fragment thereof can, for example, serve to recruit conjugated drugs, and/or can form a bispecific antibody with another monoclonal antibody to mediate the death of the targeted cancer cell.
  • the functional activity of antibodies and antigen-binding fragments thereof that bind CLDN18.2 can be characterized by methods known in the art and as described herein.
  • Methods for characterizing antibodies and antigen-binding fragments thereof that bind CLDN18.2 include, but are not limited to, affinity and specificity assays including Biacore, ELISA, and OctetRed analysis, and detection of the binding of antibodies and antigen-binding fragments to CLDN18.2 on cells (either cells transfected with CLDN18.2 or cells that naturally express CLDN18.2) by FACS. According to particular
  • the methods for characterizing antibodies and antigen-binding fragments thereof that bind CLDN18.2 include those described below.
  • the invention in another general aspect, relates to a method of treating a cancer in a subject in need thereof, comprising administering to the subject an isolated monoclonal antibody or antigen binding fragment thereof that specifically binds
  • the cancer can, for example, be selected from but not limited to, a lung cancer, a gastric cancer, an esophageal cancer, a bile duct cancer, a cholangiocarcinoma, a colon cancer, a hepatocellular carcinoma, a renal cell carcinoma, a bladder urothelial carcinoma, a metastatic melanoma, a breast cancer, an ovarian cancer, a cervical cancer, a head and neck cancer, a pancreatic cancer, a glioma, a glioblastoma, and other solid tumors, and a non-Hodgkin’s lymphoma (NHL), an acute lymphocytic leukemia (ALL), a chronic lymphocytic leukemia (CLL), a chronic myelogenous leukemia (CML), a multiple myeloma (MM), an acute myeloid leukemia (AML), and other liquid tumors.
  • NHL lymphoma
  • ALL acute lympho
  • the invention in another general aspect, relates to a method of treating an inflammatory disease in a subject in need thereof, comprising administering to the subject an isolated monoclonal antibody or antigen binding fragment thereof that specifically binds CLDN18.2 or a pharmaceutical composition of the invention.
  • the pharmaceutical composition comprises a therapeutically effective amount of an anti-CLDNl8.2 antibody or antigen binding fragment thereof.
  • therapeutically effective amount refers to an amount of an active ingredient or component that elicits the desired biological or medicinal response in a subject.
  • a therapeutically effective amount can be determined empirically and in a routine manner, in relation to the stated purpose.
  • a therapeutically effective amount means an amount of the anti- CLDN18.2 antibody or anti gen -binding fragment thereof that modulates an immune response in a subject in need thereof. Also, as used herein with reference to anti- CLDN18.2 antibodies or antigen-binding fragments thereof, a therapeutically effective amount means an amount of the anti -CLDN18.2 antibody or antigen-binding fragment thereof that results in treatment of a disease, disorder, or condition; prevents or slows the progression of the disease, disorder, or condition; or reduces or completely alleviates symptoms associated with the disease, disorder, or condition.
  • the disease, disorder or condition to be treated is cancer, preferably a cancer selected from the group consisting of a lung cancer, a gastric cancer, an esophageal cancer, a bile duct cancer, a cholangiocarcinoma, a colon cancer, a hepatocellular carcinoma, a renal cell carcinoma, a bladder urothelial carcinoma, a metastatic melanoma, a breast cancer, an ovarian cancer, a cervical cancer, a head and neck cancer, a pancreatic cancer, a glioma, a glioblastoma, and other solid tumors, and a non-Hodgkin’s lymphoma (NHL), an acute lymphocytic leukemia (ALL), a chronic lymphocytic leukemia (CLL), a chronic myelogenous leukemia (CML), a multiple myeloma (MM), an acute myeloid leukemia (AML), and
  • NHL non-Hodgkin
  • a therapeutically effective amount refers to the amount of therapy which is sufficient to achieve one, two, three, four, or more of the following effects: (i) reduce or ameliorate the severity of the disease, disorder or condition to be treated or a symptom associated therewith; (ii) reduce the duration of the disease, disorder or condition to be treated, or a symptom associated therewith; (iii) prevent the progression of the disease, disorder or condition to be treated, or a symptom associated therewith; (iv) cause regression of the disease, disorder or condition to be treated, or a symptom associated therewith; (v) prevent the development or onset of the disease, disorder or condition to be treated, or a symptom associated therewith; (vi) prevent the recurrence of the disease, disorder or condition to be treated, or a symptom associated therewith; (vii) reduce hospitalization of a subject having the disease, disorder or condition to be treated, or a symptom associated therewith; (viii) reduce
  • hospitalization length of a subject having the disease, disorder or condition to be treated, or a symptom associated therewith (ix) increase the survival of a subject with the disease, disorder or condition to be treated, or a symptom associated therewith; (xi) inhibit or reduce the disease, disorder or condition to be treated, or a symptom associated therewith in a subject; and/or (xii) enhance or improve the prophylactic or therapeutic effect(s) of another therapy.
  • the therapeutically effective amount or dosage can vary according to various factors, such as the disease, disorder or condition to be treated, the means of
  • Treatment dosages are optimally titrated to optimize safety and efficacy.
  • compositions described herein are formulated to be suitable for the intended route of administration to a subject.
  • the compositions described herein can be formulated to be suitable for intravenous, subcutaneous, or intramuscular administration.
  • the terms“treat,”“treating,” and“treatment” are all intended to refer to an amelioration or reversal of at least one measurable physical parameter related to a cancer and/or an inflammatory disease, disorder or condition, which is not necessarily discernible in the subject, but can be discernible in the subject.
  • the terms “treat,”“treating,” and“treatment,” can also refer to causing regression, preventing the progression, or at least slowing down the progression of the disease, disorder, or condition.
  • “treat,”“treating,” and“treatment” refer to an alleviation, prevention of the development or onset, or reduction in the duration of one or more symptoms associated with the disease, disorder, or condition, such as a tumor or more preferably a cancer.
  • “treat,”“treating,” and “treatment” refer to an alleviation, prevention of the development or onset, or reduction in the duration of one or more symptoms associated with the disease, disorder, or condition, such as a tumor or more preferably a cancer.
  • treatment refers to prevention of the recurrence of the disease, disorder, or condition.
  • “treat,”“treating,” and“treatment” refer to an increase in the survival of a subject having the disease, disorder, or condition.
  • “treat,”“treating,” and“treatment” refer to an increase in the survival of a subject having the disease, disorder, or condition.
  • “treat,”“treating,” and“treatment” refer to elimination of the disease, disorder, or condition in the subject.
  • compositions used in the treatment of a cancer and/or an inflammatory disease, disorder or condition can be used in combination with another treatment including, but not limited to, a chemotherapy, an anti-CD20 mAh, an anti-TIM-3 mAh, an anti -LAG-3 mAh, an anti-EGFR mAh, an anti-HER-2 mAh, an anti-CD 19 mAh, an anti-CD33 mAh, an anti-CD47 mAh, an anti-CD73 mAh, an anti -DLL-3 mAh, an anti- apelin mAh, an anti-TIP-l mAh, an anti-FOLRl mAh, an anti-CTLA-4 mAh, an anti-PD- Ll mAh, an anti-PD-l mAh, other immuno-oncology drugs, an anti angiogenic agent, a radiation therapy, an antibody-drug conjugate (ADC), a targeted therapy, or other anticancer drugs.
  • ADC antibody-drug conjugate
  • Anti-CLDNl8.2 antibodies can be used to construct bispecific antibodies with partner mAbs against PD-l, PD-L1, LAG3, TIM-3, CTLA-4, EGFR, HER-2, CD 19, CD20, CD33, CD73, CD47, CD3, apelin, DLL-3, TIP-l, folate receptor alpha (FOLR1), and/or other tumor surface antigens to treat cancers/tumors that express both CLDN18.2 and the specific tumor associated antigen.
  • partner mAbs against PD-l, PD-L1, LAG3, TIM-3, CTLA-4, EGFR, HER-2, CD 19, CD20, CD33, CD73, CD47, CD3, apelin, DLL-3, TIP-l, folate receptor alpha (FOLR1), and/or other tumor surface antigens to treat cancers/tumors that express both CLDN18.2 and the specific tumor associated antigen.
  • the term“in combination,” in the context of the administration of two or more therapies to a subject, refers to the use of more than one therapy.
  • the use of the term“in combination” does not restrict the order in which therapies are
  • a first therapy e.g., a composition described herein
  • a first therapy can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 16 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 16 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapy to a subject.
  • the invention in another general aspect, relates to a method of determining a level of CLDN18.2 in a subject.
  • the methods comprise (a) obtaining a sample from the subject; (b) contacting the sample with a monoclonal antibody or antigen -binding fragment thereof of the invention; and (c) determining a level of CLDN18.2 in the subject.
  • sample refers to a biological sample isolated from a subject and can include, but is not limited to, whole blood, serum, plasma, blood cells, endothelial cells, tissue biopsies (e.g., a cancer tissue), lymphatic fluid, ascites fluid, interstitial fluid, bone marrow, cerebrospinal fluid, saliva, mucous, sputum, sweat, urine, or any other secretion, excretion, or other bodily fluids.
  • tissue biopsies e.g., a cancer tissue
  • lymphatic fluid ascites fluid
  • interstitial fluid e.g., interstitial fluid
  • bone marrow e.g., a cancer tissue
  • cerebrospinal fluid e.g., saliva, mucous, sputum, sweat, urine, or any other secretion, excretion, or other bodily fluids.
  • A“blood sample” refers to whole blood or any fraction thereof, including blood cells, serum, and plasma.
  • the level of CLDN18.2 in the subject can be determined utilizing assays selected from, but not limited to, a Western blot assay, immunohistochemistry (IHC) and an ELISA assay. Relative protein levels can be determined by utilizing Western blot analysis and IHC, and absolute protein levels can be determined by utilizing an ELISA assay.
  • the levels of CLDN18.2 can be determined between at least two samples, e.g., between samples from the same subject at different time points, between samples from different tissues in the same subject, and/or between samples from different subjects.
  • the absolute level of CLDN18.2 in the sample can be determined by creating a standard for the ELISA assay prior to testing the sample.
  • analytical techniques to utilize to determine the level of CLDN18.2 in a sample from the subject utilizing the antibodies or anti gen -binding fragments thereof of the invention would understand which analytical techniques to utilize to determine the level of CLDN18.2 in a sample from the subject utilizing the antibodies or anti gen -binding fragments thereof of the invention.
  • CLDN18.2 levels in a disease and making appropriate therapeutic decisions.
  • a disease can be selected from, but not limited to, a cancer and an inflammatory disease.
  • the risk of developing a disease as indicated above can be determined based on the knowledge of the level of CLDN18.2 in a particular disease and/or during the progression of the particular disease.
  • Embodiment 1 is an isolated monoclonal antibody or antigen-binding fragment thereof comprising a heavy chain complementarity determining region 1 (HCDR1), HCDR2, HCDR3, a light chain complementarity determining region 1 (LCDR1),
  • HCDR1 heavy chain complementarity determining region 1
  • HCDR2 heavy chain complementarity determining region 1
  • LCDR3 light chain complementarity determining region 1
  • the antibody or antigen-binding fragment thereof specifically binds claudin 18.2 (CLDN18.2), preferably specifically binds human CLDN18.2.
  • Embodiment 2 is an isolated monoclonal antibody or antigen-binding fragment thereof comprising a heavy chain complementarity determining region 1 (HCDR1), HCDR2, HCDR3, a light chain complementarity determining region 1 (LCDR1),
  • HCDR1 heavy chain complementarity determining region 1
  • HCDR2 heavy chain complementarity determining region 1
  • LCDR3 light chain complementarity determining region 1
  • the antibody or antigen-binding fragment thereof specifically binds claudin 18.2 (CLDN18.2), preferably specifically binds human CLDN18.2.
  • Embodiment 3 is the isolated monoclonal antibody or antigen-binding fragment of embodiment 1 or 2, comprising a heavy chain variable region having a polypeptide sequence at least 95% identical to SEQ ID NO: 9, 1, 3, 5, 7, 11, 13, 15, 17 or 19, or a light chain variable region having a polypeptide sequence at least 95% identical to SEQ ID NO: 10, 2, 4, 6, 8, 12, 14, 16, 18 or 20.
  • Embodiment 4 is the isolated monoclonal antibody or antigen-binding fragment of embodiment 1 or 2, comprising
  • Embodiment 5 is the isolated monoclonal antibody or antigen-binding fragment thereof of any one of embodiments 1-4, wherein the monoclonal antibody or antigen-binding fragment thereof is capable of inducing effector-mediated tumor cell lysis.
  • Embodiment 6 is the isolated monoclonal antibody or antigen-binding fragment of any one of embodiments 1-5, wherein the antibody or antigen-binding fragment thereof is chimeric.
  • Embodiment 7 is the isolated monoclonal antibody or antigen-binding fragment of any one of embodiments 1-6, wherein the antibody or antigen -binding fragment thereof is human or humanized.
  • Embodiment 8 is the isolated monoclonal antibody or antigen-binding fragment of embodiment 7, wherein the isolated humanized antibody or antigen-binding fragment thereof comprises:
  • Embodiment 9 is the isolated monoclonal antibody or antigen-binding fragment of any one of embodiments 1-8, wherein the isolated humanized antibody or antigen-binding fragment thereof is capable of inducing effector-mediated tumor cell lysis through antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent phagocytosis (ADPC), and/or complement-dependent cytotoxicity (CDC), and/or mediating the recruitment of conjugated drugs, and/or forming a bispecific antibody with another mAh or antigen-binding fragment thereof with cancer-killing effect.
  • ADCC antibody-dependent cellular cytotoxicity
  • ADPC antibody-dependent phagocytosis
  • CDC complement-dependent cytotoxicity
  • Embodiment 10 is an isolated nucleic acid encoding the monoclonal antibody or antigen-binding fragment of any one of embodiments 1-9.
  • Embodiment 11 is a vector comprising the isolated nucleic acid of embodiment 10
  • Embodiment 12 is a host cell comprising the vector of embodiment 11.
  • Embodiment 13 is a pharmaceutical composition, comprising the isolated monoclonal antibody or anti gen -binding fragment of any one of embodiments 1-9 and a pharmaceutically acceptable carrier.
  • Embodiment 14 is a method of targeting CLDN18.2 on a cancer cell surface in a subject in need thereof, comprising administering to the subject the pharmaceutical composition of embodiment 13.
  • Embodiment 15 is a method of treating cancer in a subject in need thereof, comprising administering to the subject the pharmaceutical composition of embodiment 13.
  • Embodiment 16 is the method of embodiment 15, wherein the cancer is selected from the group consisting of a lung cancer, a gastric cancer, an esophageal cancer, a bile duct cancer, a cholangiocarcinoma, a colon cancer, a hepatocellular carcinoma, a renal cell carcinoma, a bladder urothelial carcinoma, a metastatic melanoma, a breast cancer, an ovarian cancer, a cervical cancer, a head and neck cancer, a pancreatic cancer, a glioma, a glioblastoma, and other solid tumors, and a hon- Hodgkin’s lymphoma (NHL), an acute lymphocytic leukemia (ALL), a chronic lymphocytic leukemia (CLL), a chronic myelogenous leukemia (CML), a multiple myeloma (MM), an acute myeloid leukemia (AML), and other liquid tumors.
  • NHL lymph
  • Embodiment 17 is a method of treating an inflammatory disease in a subject in need thereof, comprising administering to the subject the pharmaceutical composition of embodiment 13.
  • Embodiment 18 is a method of producing the monoclonal antibody or antigen binding fragment of any one of embodiments 1-9, comprising culturing a cell comprising a nucleic acid encoding the monoclonal antibody or antigen-binding fragment thereof under conditions to produce the monoclonal antibody or antigen-binding fragment thereof and recovering the antibody or antigen-binding fragment thereof from the cell or culture.
  • Embodiment 19 is a method of producing a pharmaceutical composition comprising the monoclonal antibody or antigen-binding fragment thereof of any one of embodiments 1-9, comprising combining the monoclonal antibody or antigen -binding fragment thereof with a pharmaceutically acceptable carrier to obtain the pharmaceutical composition.
  • Embodiment 20 is a method of determining a level of CLDN18.2 in a subject, the method comprising:
  • Embodiment 21 is the method of embodiment 20, wherein the sample is a tissue sample.
  • Embodiment 22 is the method of embodiment 21, wherein the tissue sample is a cancer tissue sample.
  • Embodiment 23 is the method of embodiment 20, wherein the sample is a blood sample.
  • mice were immunized with cells expressing exogenous human CLDN18.2 and plasma titer was determined by fluorescence-activated cell sorting (FACS). Splenocytes were harvested and fused with a myeloma cell line to produce hybridomas. Hybridomas were plated into 384 well plates and supernatants from individual wells were screened by FACS using HEK293 cells expressing CLDN18.2. Positive clones were counter-screened against human CLDN18.1 expressed on HEK293 cells by FACS. Top positive clones were isolated and sequenced.
  • FACS fluorescence-activated cell sorting
  • VH heavy chain variable region
  • VL light chain variable region
  • Table 3 CDR regions 1-3 of heavy chain for anti-CLDNl8.2 mAbs
  • HC heavy chain
  • CDR complementarity determining region
  • ID SEQ ID NO
  • the HC CDRs for the anti -CLDN18.2 mAbs were determined utilizing the IMGT method (Lefranc, M.-P. et al., Nucleic Acids Res. 1999; 27:209-212).
  • LC light chain
  • CDR complementarity determining region
  • NO SEQ ID NO
  • the LC CDRs for the anti-CLDNl8.2 mAbs were determined utilizing the IMGT method (Lefranc, M.-P. et al., Nucleic Acids Res. 1999; 27:209-212).
  • HC heavy chain
  • CDR complementarity determining region
  • N O SEQ ID NO
  • the HC CDRs for the anti -CLDN18.2 mAbs were determined utilizing a combination of IMGT (Lefranc, M.-P. et al., Nucleic Acids Res. 1999; 27:209-212) and Rabat (Elvin A. Rabat et al, Sequences of Proteins of Immunological Interest 5th ed. (1991)) methods.
  • LC light chain
  • CDR complementarity determining region
  • NO SEQ ID NO
  • the LC CDRs for the anti-CLDNl8.2 mAbs were determined utilizing a combination of IMGT (Lefranc, M.-P. et al., Nucleic Acids Res. 1999; 27:209-212) and Rabat (Elvin A. Rabat et al, Sequences of Proteins of Immunological Interest 5th ed. (1991)) methods.
  • Example 2 Production and purification of mAbs from culture media of transfected 293E cells
  • the expression vectors containing the mouse variable regions (VH and VL) fused to the constant regions of human IgGl heavy chain and kappa light chain, respectively, were transiently transfected into 293E cells.
  • the recombinant antibodies produced in the suspension of the 293E cells were purified using Protein A affinity chromatography.
  • Example 3 FACS binding analysis of purified anti-CLDN18.2 antibodies
  • HER293 cells stably transfected with full-length human CLDN18.2 were transferred to a 96-well plate. Around 50,000 cells were incubated with purified chimeric anti-CLDNl8.2 mAbs (variable regions of mouse mAbs fused to the constant regions of human IgGl heavy chain and kappa light chain, respectively) at various concentrations for 15 minutes on ice. In some instances, 100,000 cells per well were used. Cells were then centrifuged for 5 minutes and washed once with FACS buffer (HBSS supplemented with 0.1% BSA and 0.05% sodium azide).
  • FACS buffer HBSS supplemented with 0.1% BSA and 0.05% sodium azide
  • the cells were then incubated with FITC- conjugated goat anti-human IgG polyclonal antibodies (Thermo Fisher, Cat #: H10301) and incubated on ice for another 15 minutes. Cells were then washed with FACS buffer once and resuspended in FACS buffer. Cells were then run through the Attune NxT and the data were analyzed by the Attune NxT software. The FACS data are expressed as mean fluorescence intensity (MFI).
  • MFI mean fluorescence intensity
  • the chimeric anti-CLDNl8.2 mAbs were also analyzed with a HEK293 stable cell line expressing the full-length human CLDN18.2 (HEK293-CLDN18.2) using FACS as described above except that the final detection step was carried out using F(ab')2-Goat anti-Human IgG Fc conjugated to Alexa Fluor® 488 (Invitrogen, Cat#: H10120). Results for the FACS analysis of dose-dependent binding of the chimeric anti-CLDNl8.2 mAbs to the HEK293-CLDN18.2 stable cell line are provided in FIGs. 3A-3C.
  • FIGs. 4A-4J The histogram for the binding of each chimeric mAb at 7.6 nM to the HEK293-CLDN18.2 stable cell line is shown in FIGs. 4A-4J. These data indicate that the chimeric mAbs specifically bind to the HEK293-CLDN18.2 stable cell line.
  • the mouse anti- CLDN18.2 mAbs 2-C3, 5-E22, 6-J11, 3-E21 and 3-P21 were humanized to reduce the potential of immunogenicity when used in human patients.
  • the sequences of the variable regions of the heavy and light chains (VH and VL) were compared with the human antibody sequences in the Protein Data Bank (PDB) database and homology models were built.
  • the CDRs in both the heavy and light chains of the mouse mAbs were grafted into human frameworks that have the highest possibility of maintaining the proper structure likely required for antigen binding. Backmutations from human residues to mouse residue or other mutations were designed when necessary.
  • the sequences of the humanized VH and VL regions are shown in Table 7.
  • the humanized VH and VL regions were fused to the constant regions of human IgGl heavy chain and kappa light chain, respectively. Constructs corresponding to the mAh sequences were used for transient transfection in 293E or CHO cells and purified mAbs were analyzed for their ability to bind to HEK293 cells stably transfected with full-length human CLDN18.2 using FACS.
  • the ECso values of humanized mAbs for CLDN18.2 binding using a HEK293-CLDN18.2 stable pool are summarized in Table 8.
  • FIGs. 5A-5G Results for the dose-dependent binding of humanized mAbs to the HEK293- CLDN18.2 stable pool are shown in FIGs. 5A-5G.
  • FITC-based detection was used in the FACS experiments in Table 8 and FIGs 5A-5C; Alexa Fluor® 488-based detection was used in the FACS experiments in FIGs. 5D-5G.
  • FIGs. 6A-6E Results for dose-dependent binding of humanized anti-CLDNl8.2 mAbs to a HEK293-CLDN18.2 stable cell line by FACS analysis are shown in FIGs. 6A-6E.
  • the results for MJGC-4 binding by the humanized mAbs are shown in FIG 7. Alexa Fluor® 488-based detection was used in the experiments in FIGs. 6A-6E and 7.
  • ADCC antibody-dependent cellular cytotoxicity
  • Table 7 Sequences of heavy chain and light chain variable regions of humanized anti- CLDN18.2 mAbs
  • Table 8 EC50 values of humanized mAbs for CLDN18.2 binding using a HEK293- CLDN18.2 stable pool
  • 2-C3-H1L1 refers to the mAb with the 2-C3-H1 heavy chain variable region and the 2-

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Oncology (AREA)
  • Microbiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pathology (AREA)
  • Biotechnology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Hospice & Palliative Care (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

L'invention concerne des anticorps anti-CLDN18.2 et des fragments de liaison à l'antigène de ceux-ci. L'invention concerne également des acides nucléiques codant pour les anticorps, des compositions comprenant les anticorps, et des procédés de production des anticorps ainsi que l'utilisation des anticorps pour traiter ou prévenir des maladies telles que le cancer et/ou une maladie inflammatoire.
EP19764603.7A 2018-03-08 2019-03-06 Anticorps anti-claudine 18.2 et leurs utilisations Pending EP3762031A4 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201862640288P 2018-03-08 2018-03-08
US201862714254P 2018-08-03 2018-08-03
PCT/US2019/020872 WO2019173420A1 (fr) 2018-03-08 2019-03-06 Anticorps anti-claudine 18.2 et leurs utilisations

Publications (2)

Publication Number Publication Date
EP3762031A1 true EP3762031A1 (fr) 2021-01-13
EP3762031A4 EP3762031A4 (fr) 2021-12-22

Family

ID=67847436

Family Applications (1)

Application Number Title Priority Date Filing Date
EP19764603.7A Pending EP3762031A4 (fr) 2018-03-08 2019-03-06 Anticorps anti-claudine 18.2 et leurs utilisations

Country Status (13)

Country Link
US (1) US11555070B2 (fr)
EP (1) EP3762031A4 (fr)
JP (1) JP7398380B2 (fr)
KR (1) KR20200129116A (fr)
CN (1) CN111836644A (fr)
AU (1) AU2019232762B2 (fr)
BR (1) BR112020015479A2 (fr)
CA (1) CA3089653A1 (fr)
IL (1) IL276830A (fr)
MX (1) MX2020009326A (fr)
SG (1) SG11202007055QA (fr)
WO (1) WO2019173420A1 (fr)
ZA (1) ZA202004730B (fr)

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020160560A2 (fr) 2019-02-01 2020-08-06 Novarock Biotherapeutics, Ltd. Anticorps anti-claudine 18 et leurs méthodes d'utilisation
SG11202109052YA (en) * 2019-03-29 2021-10-28 Phanes Therapeutics Inc Humanized anti-claudin 18.2 chimeric antigen receptors and uses thereof
WO2021003082A1 (fr) * 2019-07-03 2021-01-07 Phanes Therapeutics, Inc. Anticorps bispecifiques anti-claudine 18.2/antic-cd47 et leurs utilisations
CN114981303B (zh) * 2019-09-13 2024-01-23 安徽俊义医疗管理咨询有限公司 人源化抗Claudin18.2(CLDN18.2)抗体
CN112707965A (zh) * 2019-09-30 2021-04-27 和铂医药(苏州)有限公司 靶向cldn18.2的抗体及其制备方法和应用
WO2021111003A1 (fr) 2019-12-06 2021-06-10 SOTIO a.s. Anticorps humanisés anti-cldn18.2
WO2021130291A1 (fr) 2019-12-23 2021-07-01 SOTIO a.s. Anticorps anti-claudin 18.2 spécifiques d'une tumeur
AU2021204861A1 (en) * 2020-01-03 2022-07-28 Crage Medical Co., Limited Anti-claudin 18.2 antibody and use thereof
CN114981308A (zh) * 2020-01-19 2022-08-30 南京金斯瑞生物科技有限公司 多特异性抗密蛋白-18.2构建体及其用途
JP2023513400A (ja) * 2020-02-10 2023-03-30 上海詩健生物科技有限公司 Cldn18.2抗体及びその使用
CN115461373B (zh) * 2020-02-10 2024-03-29 上海诗健生物科技有限公司 密蛋白18.2的抗体及其用途
WO2021257822A2 (fr) * 2020-06-18 2021-12-23 Sparx Therapeutics, Inc. Anticorps bispécifiques ciblant la claudine 18.2 humaine et le ligand de mort programmée 1 (pd-l1)
WO2022063272A1 (fr) * 2020-09-28 2022-03-31 Elpiscience (Suzhou) Biopharma, Ltd. Nouveaux anticorps anti-claudin18
CN112480248B (zh) * 2020-11-24 2023-05-05 三优生物医药(上海)有限公司 与cld18a2特异性结合的分子
WO2022122709A1 (fr) 2020-12-07 2022-06-16 Sotio Biotech A.S. Conjugués anticorps-médicament à base d'anticorps cldn18.2 humanisés
TW202241519A (zh) 2020-12-23 2022-11-01 捷克商舒迪安生物技術公司 腫瘤特異性密連蛋白18﹒2抗體藥物結合物
CN113321730B (zh) * 2021-01-11 2023-10-10 上海莱馥医疗科技有限公司 Cldn18.2抗体及其应用
CN114805571B (zh) * 2021-01-28 2023-08-25 广西鹭港生物医药科技有限公司 抗cldn18.2抗体及其应用
CN112940124B (zh) * 2021-04-14 2022-04-05 南京凯地医疗技术有限公司 靶向Claudin18.2的人源化单克隆抗体及其制备方法和应用
CN112979817B (zh) * 2021-04-27 2021-08-13 宝船生物医药科技(上海)有限公司 一种识别抗cldn18_2抗体的单克隆抗体及其制备方法和应用
CA3220892A1 (fr) * 2021-05-31 2022-12-08 Zhenhua Jia Anticorps monoclonaux contre cldn18.2 et versions modifiees par fc de ceux-ci
CN116102649A (zh) * 2021-11-11 2023-05-12 上海生物制品研究所有限责任公司 抗cldn18.2单克隆抗体及其应用
WO2023240086A1 (fr) * 2022-06-07 2023-12-14 Unm Rainforest Innovations Composition non opioïde humanisée et thérapies pour la gestion de la douleur
CN117229398A (zh) * 2022-06-15 2023-12-15 中山康方生物医药有限公司 抗cldn18.2抗体、其药物组合物及用途
WO2024002257A1 (fr) * 2022-06-30 2024-01-04 Suzhou Transcenta Therapeutics Co., Ltd. Formulation pharmaceutique stable comprenant un anticorps anti-cldn18.2
CN117777288A (zh) * 2022-09-29 2024-03-29 南京博望医药科技有限公司 抗密蛋白18.2的抗体

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUE026914T2 (en) * 2002-11-07 2016-08-29 Immunogen Inc Anti-CD33 antibodies and a method of treating acute myeloid leukemia
EP1790664A1 (fr) * 2005-11-24 2007-05-30 Ganymed Pharmaceuticals AG Anticorps monoclonaux contre claudin-18 pour le traitement du cancer
EP1914242A1 (fr) * 2006-10-19 2008-04-23 Sanofi-Aventis Nouveau anticorps Anti-CD38 pour le traitement du cancer
EP1997832A1 (fr) 2007-05-29 2008-12-03 Ganymed Pharmaceuticals AG Anticorps monoclonaux contre la claudine-18 pour le traitement contre le cancer
US8613924B2 (en) * 2009-08-06 2013-12-24 Immunas Pharma, Inc. Antibodies that specifically bind to A beta oligomers and use thereof
JP6199036B2 (ja) 2010-03-16 2017-09-20 バイオエヌテック アーゲーBioNTech AG 自己タンパク質に対する体液性免疫応答に関わる腫瘍ワクチン接種
WO2013167153A1 (fr) 2012-05-09 2013-11-14 Ganymed Pharmaceuticals Ag Anticorps utiles dans le diagnostic du cancer
WO2013174403A1 (fr) * 2012-05-23 2013-11-28 Ganymed Pharmaceuticals Ag Polythérapie impliquant des anticorps dirigés contre la claudine 18,2 pour le traitement du cancer
PL2852408T3 (pl) * 2012-05-23 2017-12-29 Ganymed Pharmaceuticals Gmbh Terapia skojarzona obejmująca przeciwciała przeciwko Klaudynie 18.2 do leczenia nowotworu
WO2013174404A1 (fr) 2012-05-23 2013-11-28 Ganymed Pharmaceuticals Ag Polythérapie impliquant des anticorps dirigés contre la claudine 18,2 pour le traitement du cancer
WO2014075697A1 (fr) 2012-11-13 2014-05-22 Biontech Ag Agents de traitement de maladies cancéreuses exprimant claudine
JP6499079B2 (ja) * 2012-11-13 2019-04-10 バイオエヌテック アーゲーBioNTech AG クローディンを発現するガン疾患を処置するための剤
EP4177273A1 (fr) * 2013-02-20 2023-05-10 Astellas Pharma Inc. Polythérapie impliquant des anticorps contre la claudine 18.2 pour le traitement du cancer
WO2014127785A1 (fr) 2013-02-20 2014-08-28 Ganymed Pharmaceuticals Ag Polythérapie impliquant des anticorps dirigés contre la claudine 18,2 pour le traitement du cancer
WO2014146672A1 (fr) 2013-03-18 2014-09-25 Ganymed Pharmaceuticals Ag Thérapie comprenant des anticorps dirigés contre cldn 18.2 pour le traitement du cancer
EP2976360B1 (fr) * 2013-03-18 2019-11-27 Astellas Pharma Inc. Therapie avec des anticorps contre claudin 18.2 pour le traitement du cancer
ES2699753T3 (es) 2014-01-29 2019-02-12 Biontech Ag Mimótopos peptídicos de claudina 18.2 y sus usos
RS61678B1 (sr) * 2014-05-28 2021-05-31 Agenus Inc Anti-gitr antitela i postupci za njihovu primenu
KR102564384B1 (ko) * 2014-07-10 2023-08-07 바이오악틱 에이비 개선된 Aβ 프로토피브릴 결합 항체
MA40871A (fr) * 2014-10-29 2017-09-05 Novartis Ag Expression directe d'anticorps
WO2016165762A1 (fr) 2015-04-15 2016-10-20 Ganymed Pharmaceuticals Ag Conjugués de médicaments comprenant des anticorps contre la claudine 18.2
WO2016165765A1 (fr) 2015-04-15 2016-10-20 Ganymed Pharmaceuticals Ag Procédés et compositions permettant de prédire l'efficacité thérapeutique des traitements contre le cancer et de pronostiquer un cancer
WO2016180468A1 (fr) * 2015-05-11 2016-11-17 Biontech Cell & Gene Therapies Gmbh Épitopes des cellules t et immunorécepteurs spécifiques de claudine-18.2
WO2016180467A1 (fr) * 2015-05-11 2016-11-17 Biontech Cell & Gene Therapies Gmbh Renforcement de l'effet de lymphocytes t génétiquement modifiés par car, par vaccination par acide nucléique
BR112018070998A2 (pt) * 2016-04-13 2019-02-26 Sanofi proteínas de ligação triespecíficas e/ou trivalentes
BR112019000327A8 (pt) * 2016-07-08 2022-10-18 Carsgen Therapeutics Co Ltd Anticorpo para anticlaudina 18a2 e uso do mesmo

Also Published As

Publication number Publication date
AU2019232762A1 (en) 2020-08-27
SG11202007055QA (en) 2020-09-29
CA3089653A1 (fr) 2019-09-12
AU2019232762B2 (en) 2023-11-16
ZA202004730B (en) 2024-01-31
MX2020009326A (es) 2020-10-08
BR112020015479A2 (pt) 2020-12-08
WO2019173420A1 (fr) 2019-09-12
JP2021514664A (ja) 2021-06-17
IL276830A (en) 2020-10-29
CN111836644A (zh) 2020-10-27
KR20200129116A (ko) 2020-11-17
US11555070B2 (en) 2023-01-17
JP7398380B2 (ja) 2023-12-14
US20200399364A1 (en) 2020-12-24
EP3762031A4 (fr) 2021-12-22

Similar Documents

Publication Publication Date Title
AU2019232762B2 (en) Anti-claudin 18.2 antibodies and uses thereof
US11401329B2 (en) Anti-CD47 antibodies and uses thereof
AU2019231172B2 (en) Anti-CD73 antibodies and uses thereof
US20210047399A1 (en) Anti-dll3 antibodies and uses thereof
US11214615B2 (en) Anti-TIM-3 antibodies and uses thereof
US11873335B2 (en) Anti-folate receptor 1 antibodies and uses thereof
WO2021003082A1 (fr) Anticorps bispecifiques anti-claudine 18.2/antic-cd47 et leurs utilisations
US11390679B2 (en) Anti-LAG-3 antibodies and uses thereof
US11306151B2 (en) Anti-TIP-1 antibodies and uses thereof
EA041922B1 (ru) Антитела против клаудина 18.2 и их применения
EA046422B1 (ru) Антитела против рецептора фолата 1 и их применения

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20201008

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

RIN1 Information on inventor provided before grant (corrected)

Inventor name: WANG, MINGHAN

Inventor name: JIA, HAIQUN

Inventor name: ZOU, HUI

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20211119

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 16/32 20060101ALI20211115BHEP

Ipc: C07K 16/30 20060101ALI20211115BHEP

Ipc: C07K 16/28 20060101ALI20211115BHEP

Ipc: A61K 39/395 20060101AFI20211115BHEP